1
|
Pan JH, Lee MK, Chang MH, Crowley LN, Le BL, Lee DS, Kim TG, Kim D, Lee K, Ko SG, Lee JH, Lee SH, Kim JK. Optimized combination of Cervus nippon (Sika deer), Angelica (Dangui), and Rehmannia (Suk-jihwang) mitigates LPS-induced inflammation: exploring signaling pathways through plasma metabolomics. Food Sci Biotechnol 2024; 33:1671-1683. [PMID: 38623429 PMCID: PMC11016027 DOI: 10.1007/s10068-023-01476-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 10/01/2023] [Accepted: 10/12/2023] [Indexed: 04/17/2024] Open
Abstract
This study aimed to determine the optimal combination of three anti-inflammatory materials [i.e., Cervus nippon Temminck (CT), Angelica gigas Nakai (AN), and Rehmannia glutinosa (RG)] for the strongest anti-inflammatory potential. Eighteen combinations of the three materials were tested in LPS-stimulated RAW264.7 cells via assessing nitric oxide (NO). The best combination from in vitro studies was administered to LPS-treated C57BL/6J mice for five days. Subsequently, plasma metabolites were profiled by bioinformatics analyses and validations. As results, 2, 20, and 50 µg/mL of CT, AN, and RG (TM) were the most effective combination suppressing inflammation. In mice, TM mitigated hepatic inflammatory markers. Similarly, the metabolomics indicated that TM may suppress NF-κB signaling pathway, thereby alleviating hepatic inflammation. TM also decreased systemic and hepatic pro-inflammatory cytokines. Collectively, we found the optimal combination of TM for mitigating inflammation; thus further studies on safety, mechanisms, and clinical models are warranted for human applications. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-023-01476-x.
Collapse
Affiliation(s)
- Jeong Hoon Pan
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE 19716 USA
- Department of Food and Nutrition, Chosun University, Gwangju, 61452 Republic of Korea
| | - Min Kook Lee
- Department of Food and Biotechnology, Korea University, 2511 Sejong-ro, Sejong, 30019 Republic of Korea
- BK21 FOUR Research Group for Omics-based Bio-health in Food Industry, Korea University, Sejong, Republic of Korea
| | - Moon Han Chang
- Department of Food and Biotechnology, Korea University, 2511 Sejong-ro, Sejong, 30019 Republic of Korea
- BK21 FOUR Research Group for Omics-based Bio-health in Food Industry, Korea University, Sejong, Republic of Korea
| | - Liana N. Crowley
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE 19716 USA
| | - Brandy L. Le
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE 19716 USA
| | - Da Seul Lee
- Agricultural Microbiology Division, National Institute of Agricultural Sciences, Rural Development Administration, Wanju, 55365 Republic of Korea
| | - Tae Gyun Kim
- The Bioinformatix Inc, Cheongju, 28674 Republic of Korea
| | - Dahye Kim
- Animal Genomics and Bioinformatics Division, National Institute of Animal Science, Rural Development Administration, Wanju, 55365 Republic of Korea
| | - Kangwook Lee
- Department of Food and Biotechnology, Korea University, 2511 Sejong-ro, Sejong, 30019 Republic of Korea
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447 Republic of Korea
| | - Seong-Gyu Ko
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447 Republic of Korea
| | - Jin Hyup Lee
- Department of Food and Biotechnology, Korea University, 2511 Sejong-ro, Sejong, 30019 Republic of Korea
- BK21 FOUR Research Group for Omics-based Bio-health in Food Industry, Korea University, Sejong, Republic of Korea
- Biological Clock-Based Anti-aging Convergence RLRC, Korea University, Sejong, Republic of Korea
| | - Suk Hee Lee
- Department of Food and Biotechnology, Korea University, 2511 Sejong-ro, Sejong, 30019 Republic of Korea
- BK21 FOUR Research Group for Omics-based Bio-health in Food Industry, Korea University, Sejong, Republic of Korea
- Biological Clock-Based Anti-aging Convergence RLRC, Korea University, Sejong, Republic of Korea
| | - Jae Kyeom Kim
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE 19716 USA
- Department of Food and Biotechnology, Korea University, 2511 Sejong-ro, Sejong, 30019 Republic of Korea
- Department of Behavioral Health and Nutrition, University of Delaware, 15 Innovation Way, Newark, DE 19711 USA
| |
Collapse
|
2
|
de Souza Sampaio R, Brito Pereira Bezerra Martins AO, Santos da Silva LY, Torres Pessoa DR, Ribeiro-Filho J, de Araújo Delmondes G, Bezerra Felipe CF, Alencar de Menezes IR, Kerntopf MR. Topical Antiedematogenic Activity of the Essential Oil of Psidium brownianum Mart. (OEPB) in Murine Ear Edema Models. Chem Biodivers 2024; 21:e202400187. [PMID: 38429232 DOI: 10.1002/cbdv.202400187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/01/2024] [Accepted: 03/01/2024] [Indexed: 03/03/2024]
Abstract
Psidium brownianum Mart is reported in the literature by antinociceptive and antioxidant activities, indicating that this species' secondary metabolites might be used to control inflammatory processes. The present study aimed to characterize the topical antiedematogenic activity of the essential oil of Psidium brownianum Mart. (OEPB) in ear edema models by different inflammatory agents. Female Swiss mice (25-35 g) and Wistar albino rats (200-300 g) were used throughout tests (n=6/group) on acute or chronic edema models induced by single and multiple topical applications. The OEPB is administered topically pure or at a concentration of 100 or 200 mg/mL. The antiedematogenic mechanism of OEPB was analyzed by administering capsaicin, arachidonic acid, histamine, and phenol at the best effective dose (200 mg/mL). The results showed a significant reduction of edema-induced single (28.87 %) and multiple (50.13 %) applications of croton oil compared to the negative control group. Regarding potential mechanisms of action, OEPB (200 mg/mL) inhibited the development of edema triggered by capsaicin (29.95 %), arachidonic acid (22.66 %), phenol (23.35 %), and histamine (75.46 %), suggesting an interference with the histaminergic pathway. These results indicate that OEPB presents a topical antiedematogenic effect in acute and chronic murine models, possibly interfering with inflammatory pathways triggered by mediators such as histamine.
Collapse
Affiliation(s)
- Renata de Souza Sampaio
- Laboratory of Pharmacology and Molecular Chemistry (LFQM), Department of Biological Chemistry, Regional University of Cariri (URCA), 63105-000, Crato, CE, Brazil
| | | | - Lucas Yure Santos da Silva
- Laboratory of Pharmacology and Molecular Chemistry (LFQM), Department of Biological Chemistry, Regional University of Cariri (URCA), 63105-000, Crato, CE, Brazil
| | - Dra Renata Torres Pessoa
- Laboratory of Pharmacology and Molecular Chemistry (LFQM), Department of Biological Chemistry, Regional University of Cariri (URCA), 63105-000, Crato, CE, Brazil
| | - Jaime Ribeiro-Filho
- Oswaldo Cruz Foundation (Fiocruz), Fiocruz Ceará, 60180-900, Eusébio, CE, Brazil
| | - Gyllyandeson de Araújo Delmondes
- Natural Products Pharmacology Laboratory, Department of Biological Chemistry, Regional University of Cariri (URCA), 63105-000, Crato, CE, Brazil
| | - Cícero Francisco Bezerra Felipe
- Molecular Biology Departament, Federal University of Paraiba (UFPB), Campus I - Jardim Cidade Universitária, 58059-900, João Pessoa, PB, Brazil
| | - Irwin Rose Alencar de Menezes
- Laboratory of Pharmacology and Molecular Chemistry (LFQM), Department of Biological Chemistry, Regional University of Cariri (URCA), 63105-000, Crato, CE, Brazil
| | - Marta Regina Kerntopf
- Laboratory of Pharmacology and Molecular Chemistry (LFQM), Department of Biological Chemistry, Regional University of Cariri (URCA), 63105-000, Crato, CE, Brazil
| |
Collapse
|
3
|
Liu Y, Meng C, Li Y, Xia D, Lu C, Lai J, Zhang Y, Cao K, Gao X, Yuan Q. Peptide-Protected Gold Nanoclusters Efficiently Ameliorate Acute Contact Dermatitis and Psoriasis via Repressing the TNF-α/NF-κB/IL-17A Axis in Keratinocytes. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:662. [PMID: 36839031 PMCID: PMC9963485 DOI: 10.3390/nano13040662] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 06/18/2023]
Abstract
Immune-mediated skin diseases have a high prevalence and seriously affect patients' quality of life. Gold compounds have been considered promising therapeutic agents in dermatology, but the high incidence of adverse reactions have limited their clinical application. There is a great need to develop more effective and less toxic gold-based drugs. Gold nanoclusters fabricated by using peptides (pep-AuNCs) have appeared as potential biomedical nanomaterials because of their excellent biocompatibility, ease of fabrication and unique physicochemical properties. Glutathione (GSH) is an endogenous tripeptide and has been used for lightening the skin color. Therefore, we fabricated a well-defined gold nanocluster with GSH as an example to explore the immunomodulatory effect of AuNCs on a TNF-α-treated human keratinocyte cell line (HaCaT) in vitro, the 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced irritant contact dermatitis (ICD) model and the oxazolone (OXA)-induced psoriatic model in vivo. The results indicated that topically applied AuNCs successfully attenuated the severity of ICD and psoriasis-like lesions. In vitro and in vivo, AuNCs effectively inhibited the abnormal activation of the NF-κB pathway and the consequent overexpression of proinflammatory cytokines in keratinocytes. In particular, the transactivation of IL-17A, the most important cytokine in psoriasis pathology, was effectively inhibited by AuNCs treatment. In addition, AuNCs did not show any obvious cytotoxicity in HaCaT cells at doses even up to 100 µM and did not induce any irritation in the healthy skin and major organs, which indicated their favorable biosafety. These results indicate that biocompatible pep-AuNCs might be a promising gold-based nanomedicine for the treatment of inflammatory skin diseases.
Collapse
Affiliation(s)
- Yu Liu
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| | - Cong Meng
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| | - Yanggege Li
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| | - Dongfang Xia
- College of Chemistry and Material Science, Shandong Agricultural University, Taian 271018, China
| | - Cao Lu
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| | - Jing Lai
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| | - Yulu Zhang
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| | - Kai Cao
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| | - Xueyun Gao
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| | - Qing Yuan
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
4
|
Chang SN, Park JG, Kang SC. Therapeutic propensity of ginsenosides Rg1 and Rg3 in rhabdomyolysis-induced acute kidney injury and renohepatic crosstalk in rats. Int Immunopharmacol 2023; 115:109602. [PMID: 36580761 DOI: 10.1016/j.intimp.2022.109602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND Ginseng is a traditional herbal medicine used for thousands of years in Southeast Asian countries because of its medicinal properties. Ginsenosides Rg1 and Rg3 have demonstrated therapeutic properties against a broad spectrum of diseases. PURPOSE Here in this study, we investigated the therapeutic efficacy of Rg1 and Rg3 in alleviating glycerol-induced acute kidney injury, also known as rhabdomyolysis-induced acute kidney injury (RAKI). METHODS AKI was induced in male Wistar rats through intramuscular injection of 10 mL/kg glycerol and simultaneous oral treatment of ginsenosides Rg1 and Rg3 for 3 days. We also evaluated the therapeutic potential of Rg1 and Rg3 on human embryonic kidney epithelial (HEK-293). Cell viability and LDH assay were performed on HEK-293 cells to evaluate the toxicity of Rg1 and Rg3. Evaluation of important kidney damage markers such as creatinine and blood urea nitrogen (BUN) was carried out at different time points from the rat serum. Histopathological analysis was performed on kidney tissues. We also performed experiments such as ELISA assay, immunohistochemistry, immunofluorescence staining, COMET assay, western blotting, TUNEL assay, and flow cytometry to obtain results. RESULTS Rg1 and Rg3 significantly downregulated the expression of kidney damage markers such as creatinine and BUN in a dose-dependent manner. Histopathological analysis revealed damage across the glomerulus, tubules, and collecting duct rendering the kidney dysfunctional in glycerol treatment groups. However, Rg1 and Rg3 treated groups showed a significant reduction in tubular necrosis at both 10 and 20 mg/kg. There was also a sharp downregulation of oxidative and ER stress markers. Additionally, we observed nuclear translocation of Nrf2 which were more prominent in kidney tissues. Rg1 and Rg3 were also able to mitigate apoptotic cell death in vitro and in vivo evaluated through immunofluorescence staining for p53, TUNEL assay, flow cytometry, and immunoblotting for intrinsic apoptosis markers. CONCLUSION In summary, we conclude that Rg1 and Rg3 exhibited natural therapeutic remedy against AKI.
Collapse
Affiliation(s)
- Sukkum Ngullie Chang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea.
| | - Jae Gyu Park
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Republic of Korea.
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea.
| |
Collapse
|
5
|
Chang SN, Keretsu S, Kang SC. Evaluation of decursin and its isomer decursinol angelate as potential inhibitors of human glutamate dehydrogenase activity through in silico and enzymatic assay screening. Comput Biol Med 2022; 151:106287. [PMID: 36455296 DOI: 10.1016/j.compbiomed.2022.106287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 10/09/2022] [Accepted: 11/06/2022] [Indexed: 11/16/2022]
Abstract
Glutaminolysis is a typical hallmark of malignant tumors across different cancers. Glutamate dehydrogenase (GDH, GLUD1) is one such enzyme involved in the conversion of glutamate to α-ketoglutarate. High levels of GDH are associated with numerous diseases and is also a prognostic marker for predicting metastasis in colorectal cancer. Therefore, inhibiting GDH can be a crucial therapeutic target. Here in this study, we performed molecular docking analysis of 8 different plants derived single compounds collected from pubChem database for screening and selected decursin (DN) and decursinol angelate (DA). We performed molecular dynamics simulation (MD), monitored the stability, interaction for protein and docked ligand at 50 ns, and evaluated the molecular mechanics Poisson-Boltzmann surface area (MM-PBSA) free energy calculation on the twoselected compounds along with a standard inhibitor epigallocatechin gallate (EGCG) as reference. The final results showed the formation of stable hydrogen bond interactions by DN and DA in the residues of R400 and Y386 at the ADP activation site of GDH, which was important for the selective inhibition of GDH activity. Additionally, the total binding energy of DN and DA were -115.5 kJ/mol and -106.2 kJ/mol, which was higher than the standard reference GDH inhibitor EGCG (-92.8 kJ/mol). Furthermore, biochemical analysis for GDH inhibition substantiated our computational results and established DN and DA as novel GDH inhibitor. The percentage of IC50 inhibition for DN and DA were 1.035 μM and 1.432 μM. Conclusively, DN and DA can be a novel therapeutic drug for inhibition of glutamate dehydrogenase.
Collapse
Affiliation(s)
| | - Seketoulie Keretsu
- Department of Pathology, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, 38453, South Korea.
| |
Collapse
|
6
|
Chang SN, Haroon M, Dey DK, Kang SC. Rhabdomyolysis-induced acute kidney injury and concomitant apoptosis induction via ROS-mediated ER stress is efficaciously counteracted by epigallocatechin gallate. J Nutr Biochem 2022; 110:109134. [PMID: 36028100 DOI: 10.1016/j.jnutbio.2022.109134] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/03/2022] [Accepted: 08/05/2022] [Indexed: 01/13/2023]
Abstract
Rhabdomyolysis induced acute kidney injury (RIAKI) is a life-threatening condition responsible for approximately 19-58% of AKI cases worldwide. We performed an intramuscular injection of glycerol (10 mL/kg) in male wistar rats to induce AKI. Epigallocatechin gallate (EGCG) was administered for 3 consecutive days to evaluate its protective effects. We observed significant downregulation in serum creatinine, blood urea nitrogen (BUN) and LDH at different time points on EGCG treatment groups in a dose-dependent manner. Similarly, H&E staining also revealed that EGCG was able to reduce the formation of damaged tubules and tubular necrosis which was prominently spread throughout the kidney tissue of glycerol treatment group. Concomitantly, we observed upregulated inflammation, ER stress and elevated oxidative stress in the glycerol treated group only, which was significantly normalized upon EGCG treatment in both in vitro and in vivo studies. The occurrence of apoptosis in kidney tubules was found to be relatively higher in glycerol treated group and H2O2 treated HEK-293 cells. The results obtained after EGCG treatment revealed a significant decrease in apoptotic cell population, which was further validated by immunofluorescence staining against p53 and comet assay in HEK-293 cells and p53 IHC in kidney tissues. Western blotting also revealed a systemic downregulation of intrinsic mitochondrial apoptotic pathway markers such as bax, bcl-2, pro and cleaved caspase 3, caspase 9 and PARP1. Additionally, the results for flow cytometry analysis and TUNEL assay corroborated apoptotic equilibrium. Conclusively, we reckon EGCG as a multi-therapeutic natural product that can be used the for treatment of AKI.
Collapse
Affiliation(s)
- Sukkum Ngullie Chang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Korea
| | - Muhammad Haroon
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Korea
| | - Debasish Kumar Dey
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Korea; Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK 74117, USA
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Korea.
| |
Collapse
|
7
|
Zhao F, Barber CJ, Sammani S, Wan L, Miller BW, Furenlid LR, Li Z, Gotur DB, Barrios R, Woolfenden JM, Martin DR, Liu Z. Use of radiolabeled hyaluronic acid for preclinical assessment of inflammatory injury and acute respiratory distress syndrome. Nucl Med Biol 2022; 114-115:86-98. [PMID: 36270074 PMCID: PMC9562607 DOI: 10.1016/j.nucmedbio.2022.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/29/2022] [Accepted: 10/06/2022] [Indexed: 12/27/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is accompanied by a dramatic increase in lung hyaluronic acid (HA), leading to a dose-dependent reduction of pulmonary oxygenation. This pattern is associated with severe infections, such as COVID-19, and other important lung injury etiologies. HA actively participates in molecular pathways involved in the cytokine storm of COVID-19-induced ARDS. The objective of this study was to evaluate an imaging approach of radiolabeled HA for assessment of dysregulated HA deposition in mouse models with skin inflammation and lipopolysaccharide (LPS)-induced ARDS using a novel portable intensified Quantum Imaging Detector (iQID) gamma camera system. METHODS HA of 10 kDa molecular weight (HA10) was radiolabeled with 125I and 99mTc respectively to produce [125I]I-HA10 and [99mTc]Tc-HA10, followed by comparative studies on stability, in vivo biodistribution, and uptake at inflammatory skin sites in mice with 12-O-tetradecanoylphorbol-13-acetate (TPA)-inflamed ears. [99mTc]Tc-HA10 was used for iQID in vivo dynamic imaging of mice with ARDS induced by intratracheal instillation of LPS. RESULTS [99mTc]Tc-HA10 and [125I]I-HA10 had similar biodistribution and localization at inflammatory sites. [99mTc]Tc-HA10 was shown to be feasible in measuring skin injury and monitoring skin wound healing. [99mTc]Tc-HA10 dynamic pulmonary images yielded good visualization of radioactive uptake in the lungs. There was significantly increased lung uptake and slower lung washout in mice with LPS-induced ARDS than in control mice. Postmortem biodistribution measurement of [99mTc]TcHA10 (%ID/g) was 11.0 ± 3.9 vs. 1.3 ± 0.3 in the ARDS mice (n = 6) and controls (n = 6) (P < 0.001), consistent with upregulated HA expression as determined by enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry (IHC) staining. CONCLUSIONS [99mTc]Tc-HA10 is promising as a biomarker for evaluating HA dysregulation that contributes to pulmonary injury in ARDS. Rapid iQID imaging of [99mTc]Tc-HA10 clearance from injured lungs may provide a functional template for timely assessment and quantitative monitoring of pulmonary pathophysiology and intervention in ARDS.
Collapse
Affiliation(s)
- Fangyuan Zhao
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America; Food Science and Engineering College, Qingdao Agricultural University, China
| | - Christy J Barber
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America
| | - Saad Sammani
- Department of Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Li Wan
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America
| | - Brian W Miller
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America; College of Optical Sciences, University of Arizona, Tucson, AZ, United States of America
| | - Lars R Furenlid
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America; College of Optical Sciences, University of Arizona, Tucson, AZ, United States of America
| | - Zheng Li
- Department of Radiology, Houston Methodist Hospital, Houston, TX, United States of America
| | - Deepa B Gotur
- Department of Medicine, Houston Methodist Hospital, Houston, TX, United States of America
| | - Roberto Barrios
- Department of Pathology, Houston Methodist Hospital, Houston, TX, United States of America
| | - James M Woolfenden
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America
| | - Diego R Martin
- Department of Radiology, Houston Methodist Hospital, Houston, TX, United States of America
| | - Zhonglin Liu
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America; Department of Radiology, Houston Methodist Hospital, Houston, TX, United States of America.
| |
Collapse
|
8
|
Albayrak G, Demir S, Koyu H, Baykan S. Anticholinesterase Compounds from Endemic Prangos uechtritzii. Chem Biodivers 2022; 19:e202200557. [PMID: 36201258 DOI: 10.1002/cbdv.202200557] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/04/2022] [Indexed: 11/10/2022]
Abstract
In this study, the anticholinesterase effects of the extracts and isolated compounds from the roots of endemic Prangos uechtritzii Boiss & Hausskn (Apiaceae) are reported. A novel polyacetylenic compound; (+)-8-O-methyloplopantriol A along with two known polyacetylenes; (-)-panaxynol, (+)-falcarindiol and fifteen known coumarin derivatives; umbelliferone, 6-formylumbelliferone, suberosin, 7-demethylsuberosin, (+)-ulopterol, tamarin, psoralen, imperatorin, (+)-oxypeucedanin, (+)-oxypeucedanin hydrate, (+)-oxypeucedanin methanolate, (+)-marmesin, (-)-prantschimgin, (+)-decursinol, and (-)-adicardin were isolated from the hexane (Pu-HE), chloroform (Pu-CE), and methanol (Pu-ME) extracts of P. uechtritzii roots. (-)-Panaxynol, (+)-falcarindiol, 6-formylumbelliferone, (+)-decursinol, and (-)-adicardin were obtained from the genus Prangos for the first time. (+)-8-O-Methyloplopantriol A inhibited both AChE (IC50 =194.5±5.8 μM) and BChE (IC50 =51.9±2.96 μM) enzymes. (+)-Falcarindiol, 6-formylumbelliferone, 7-demethylsuberosin, tamarin, and imperatorin also exhibited BChE-specific inhibitory activities (IC50 =27.88-93.86 μM). (+)-Falcarindiol (IC50 =27.88±0.91 μM) and imperatorin (IC50 =30.89±1.40 μM) as the most active components could be led compounds to develop new BChE inhibitors with further research against Alzheimer's disease.
Collapse
Affiliation(s)
- Gokay Albayrak
- Department of Pharmaceutical Botany, Faculty of Pharmacy, İzmir Katip Çelebi University, 35620, İzmir, Turkey
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Ege University, 35040, İzmir, Turkey
| | - Serdar Demir
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Ege University, 35040, İzmir, Turkey
| | - Halil Koyu
- Department of Pharmaceutical Botany, Faculty of Pharmacy, İzmir Katip Çelebi University, 35620, İzmir, Turkey
| | - Sura Baykan
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Ege University, 35040, İzmir, Turkey
| |
Collapse
|
9
|
Zhang S, Zhang L, Yu M, Luo D, Chen S, Liu W, Zhang Y, Zhang L, Zhao T. Essential oils of Zingiber officinale: Chemical composition, in vivo alleviation effects on TPA induced ear swelling in mice and in vitro bioactivities. Front Nutr 2022; 9:1043175. [PMID: 36352904 PMCID: PMC9639606 DOI: 10.3389/fnut.2022.1043175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/03/2022] [Indexed: 11/24/2022] Open
Abstract
Zingiber officinale (ZO) is a traditional food condiment. The essential oils of Z. officinale (ZOEOs) are known to have multiple bioactivities. In this study, gas chromatography mass spectrometer (GC-MS) analytical method was used to identify active ingredient present in ZOEOs. A total of 41 compounds were identified in ZOEOs. Major components in ZOEOs were zingiberene (19.71%), (+)-β-cedrene (12.85%), farnesene (12.17%), α-curcumene (10.18%) and β-elemene (3.54%). Experimental results of 12-O-tetradecanoylphorbol-13 acetate (TPA) induced ear swelling validation mice model showed that ZOEOs treatment has better anti-inflammatory effect compared with ibuprofen (positive control) at high concentrations. Histological and immunohistochemical analysis showed that ZOEOs significantly decreased COX-2, IL-6 and NF-κB expression in a dose dependent manner. The mRNA levels of COX-2 and NF-κB were also down regulated by the application of ZOEOs. This indicated that ZOEOs exhibited positive effects in ear skin protection. Antibacterial experimental results showed that EOZOs had anti-bacterial effects on Escherichia coli, Pseudomonas aeruginosa, and Staphylococcus aureus. DPPH radical scavenging, A549 cell line and LNCaP cell line inhibition results indicated that ZOEOs exhibited potential antioxidant and anti-tumor properties. The findings of these study provide scientific basis on therapeutic use of ZO in food, cosmetic and pharmaceutical industries.
Collapse
Affiliation(s)
- Silu Zhang
- Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Agricultural Products Processing, Sericulture and Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Shenzhen Precision Health Food Technology Co., Ltd., Shenzhen, China
| | - Lijun Zhang
- Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Agricultural Products Processing, Sericulture and Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Mengjie Yu
- Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Agricultural Products Processing, Sericulture and Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Donghui Luo
- Guangdong Provincial Key Laboratory of Plant Resources Biorefinery, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Shuai Chen
- Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Agricultural Products Processing, Sericulture and Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Weifeng Liu
- Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Agricultural Products Processing, Sericulture and Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Yehui Zhang
- Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Agricultural Products Processing, Sericulture and Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Lanyue Zhang
- Guangdong Provincial Key Laboratory of Plant Resources Biorefinery, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
- *Correspondence: Lanyue Zhang
| | - Tiantian Zhao
- Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Agricultural Products Processing, Sericulture and Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Food Science and Engineering Department, Chaozhou Branch of Chemistry and Chemical Engineering Guangdong Laboratory, Chaozhou, China
- Tiantian Zhao
| |
Collapse
|
10
|
Karthika C, Najda A, Klepacka J, Zehravi M, Akter R, Akhtar MF, Saleem A, Al-Shaeri M, Mondal B, Ashraf GM, Tagde P, Ramproshad S, Ahmad Z, Khan FS, Rahman MH. Involvement of Resveratrol against Brain Cancer: A Combination Strategy with a Pharmaceutical Approach. Molecules 2022; 27:4663. [PMID: 35889532 PMCID: PMC9320031 DOI: 10.3390/molecules27144663] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 11/25/2022] Open
Abstract
A brain tumor (BT) is a condition in which there is growth or uncontrolled development of the brain cells, which usually goes unrecognized or is diagnosed at the later stages. Since the mechanism behind BT is not clear, and the various physiological conditions are difficult to diagnose, the success rate of BT is not very high. This is the central issue faced during drug development and clinical trials with almost all types of neurodegenerative disorders. In the first part of this review, we focus on the concept of brain tumors, their barriers, and the types of delivery possible to target the brain cells. Although various treatment methods are available, they all have side effects or toxic effects. Hence, in the second part, a correlation was made between the use of resveratrol, a potent antioxidant, and its advantages for brain diseases. The relationship between brain disease and the blood-brain barrier, multi-drug resistance, and the use of nanomedicine for treating brain disorders is also mentioned. In short, a hypothetical concept is given with a background investigation into the use of combination therapy with resveratrol as an active ingredient, the possible drug delivery, and its formulation-based approach.
Collapse
Affiliation(s)
- Chenmala Karthika
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty 643001, India;
| | - Agnieszka Najda
- Department of Vegetable and Herbal Crops, University of Life Science in Lublin, Doświadczalna Street 51A, 20280 Lublin, Poland
| | - Joanna Klepacka
- Department of Commodity Science and Food Analysis, Faculty of Food Science, University of Warmia and Mazury in Olsztyn, Oczapowskiego 2, 10719 Olsztyn, Poland;
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy Girls Section, Prince Sattam Bin Abdul Aziz University, Alkharj 11942, Saudi Arabia;
| | - Rokeya Akter
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea;
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Lahore Campus, Riphah International University, Lahore 54950, Pakistan;
| | - Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan;
| | - Majed Al-Shaeri
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Banani Mondal
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj 1400, Bangladesh; (B.M.); (S.R.)
| | - Ghulam Md. Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Priti Tagde
- Amity Institute of Pharmacy, Amity University, Noida 201301, India;
| | - Sarker Ramproshad
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj 1400, Bangladesh; (B.M.); (S.R.)
| | - Zubair Ahmad
- Unit of Bee Research and Honey Production, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia;
- Biology Department, College of Arts and Sciences, Dehran Al-Junub, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia;
| | - Farhat S. Khan
- Biology Department, College of Arts and Sciences, Dehran Al-Junub, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia;
| | - Md. Habibur Rahman
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea;
| |
Collapse
|
11
|
Kim CG, Chang SN, Park SM, Hwang BS, Kang SA, Kim KS, Park JG. Moringa oleifera mitigates ethanol-induced oxidative stress, fatty degeneration and hepatic steatosis by promoting Nrf2 in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 100:154037. [PMID: 35358929 DOI: 10.1016/j.phymed.2022.154037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/04/2022] [Accepted: 03/09/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Moringa oleifera (M. oleifera) is cultivated throughout the world and it is known by numerous regional names and is consumed as medication for various diseases such as hypertension, diabetes, HIV and is potential source of nutrients and natural antioxidants making it among the most useful trees. METHODS We evaluated the therapeutic potential of M. oleifera on ethanol-induced fatty liver. The mice were treated with 30% ethanol (EtOH) alone or in combination with different concentration of M. oleifera extracts (100, 200 and 400 mg/kg). We performed biochemical estimation for the serum of important liver damage markers such as aspartate aminotransferase (AST), alanine aminotransferase (ALT) and triglyceride (TG). We performed histopathological analysis from the liver tissues of different mice groups. We also performed ELISA assay, western blotting analysis and SPECT imaging to obtain our results. RESULTS The results for serum (AST, p < 0.0001), (ALT, p < 0.0006) and triglyceride (TG, p < 0.0003) were found to be significantly reduced in all doses of M. oleifera extract treatment groups in comparison with the ethanol group. H&E staining analysis and scoring revealed a significant reduction in lipid droplet accumulation and a significant reduction of liver steatosis (p < 0.0001), lobular inflammation (p < 0.0013), ballooning (p < 0.0004) and immunohistochemistry for TNF-α. M. oleifera also ameliorated ethanol-induced oxidative stress evaluated through MDA (p < 0.0001), H2DCFDA, JC-1 staining and a significant down-regulation of CYP2E1 enzyme (p < 0.0001) in the 200 and 400 mg/kg groups in comparison with EtOH groups. M. oleifera extract also boosted the antioxidant response evaluated through total GSH assay (p < 0.0001) and nuclear translocation of Nrf2. Furthermore, we performed SPECT imaging and evaluated the liver uptake value (LUV) to assess the extent of liver damage. LUV was observed to be lower in the ethanol group, whereas LUV was higher in control and M. olifera treated groups. CONCLUSION In summary, from this experiment we conclude that M. oleifera extract has the potential to ameliorate ethanol-induced liver damage.
Collapse
Affiliation(s)
- Chang Geon Kim
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang, Gyeongbuk, 37668, Republic of Korea; School of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 712749, Republic of Korea
| | - Sukkum Ngullie Chang
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang, Gyeongbuk, 37668, Republic of Korea; Department of Biotechnology, Daegu University, Gyeongsan, 38453, Republic of Korea
| | - Seon Min Park
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang, Gyeongbuk, 37668, Republic of Korea; Department of Veterinary Toxicology, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea
| | - Buyng Su Hwang
- Nakdonggang National Institute of Biological Resources, Sangju, 37242, Republic of Korea
| | - Sung-A Kang
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang, Gyeongbuk, 37668, Republic of Korea
| | - Kil Soo Kim
- Department of Veterinary Toxicology, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea.
| | - Jae Gyu Park
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang, Gyeongbuk, 37668, Republic of Korea.
| |
Collapse
|
12
|
Wang XL, Jiang RW. Therapeutic Potential of Superoxide Dismutase Fused with Cell-Penetrating Peptides in Oxidative Stress-Related Diseases. Mini Rev Med Chem 2022; 22:2287-2298. [PMID: 35227183 DOI: 10.2174/1389557522666220228150127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/17/2021] [Accepted: 12/27/2021] [Indexed: 11/22/2022]
Abstract
Superoxide dismutase (SOD) is a well-known cellular antioxidant enzyme. However, exogenous SOD cannot be used to protect tissues from oxidative damage due to the low permeability of the cell membrane. Cell-penetrating peptides (CPPs) are a class of short peptides that can cross the cell membrane. Recombinant fusion protein that fuses SOD protein with CPP (CPP-SOD) can cross various tissues and organs as well as the blood-brain barrier. CPP-SODs can relieve severe oxidative damage in various tissues caused by radiation, ischemia, inflammation, and chemotherapy by clearing the reactive oxygen species, reducing the expression of inflammatory factors, and inhibiting NF-κB/MAPK signaling pathways. Therefore, the clinical application of CPP-SODs provide new therapeutic strategies for a variety of oxidative stress-related disorders, such as Parkinson's disease, diabetes, obesity, cardiac fibrosis, and premature aging.
Collapse
Affiliation(s)
- Xiao-Lu Wang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, and International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Ren-Wang Jiang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, and International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, Jinan University, Guangzhou 510632, China
| |
Collapse
|
13
|
Yeo EJ, Shin MJ, Yeo HJ, Choi YJ, Sohn EJ, Lee LR, Kwon HJ, Cha HJ, Lee SH, Lee S, Yu YH, Kim DS, Kim DW, Park J, Han KH, Eum WS, Choi SY. Tat-thioredoxin 1 reduces inflammation by inhibiting pro-inflammatory cytokines and modulating MAPK signaling. Exp Ther Med 2021; 22:1395. [PMID: 34650643 DOI: 10.3892/etm.2021.10831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/29/2021] [Indexed: 10/20/2022] Open
Abstract
Thioredoxin 1 (Trx1) serves a central role in redox homeostasis. It is involved in numerous other processes, including oxidative stress and apoptosis. However, to the best of our knowledge, the role of Trx1 in inflammation remains to be explored. The present study investigated the function and mechanism of cell permeable fused Tat-Trx1 protein in macrophages and a mouse model. Transduction levels of Tat-Trx1 were determined via western blotting. Cellular distribution of transduced Tat-Trx1 was determined by fluorescence microscopy. 2',7'-Dichlorofluorescein diacetate and TUNEL staining were performed to determine the production of reactive oxygen species and DNA fragmentation. Protein and gene expression were measured by western blotting and reverse transcription-quantitative PCR (RT-qPCR), respectively. Effects of skin inflammation were determined using hematoxylin and eosin staining, changes in ear weight and ear thickness, and RT-qPCR in ear edema animal models. Transduced Tat-Trx1 inhibited lipopolysaccharide-induced cytotoxicity and activation of NF-κB, MAPK and Akt. Additionally, Tat-Trx1 markedly reduced the production of inducible nitric oxide synthase, cyclooxygenase-2, IL-1β, IL-6 and TNF-α in macrophages. In a 12-O-tetradecanoylphorbol-13-acetate-induced mouse model, Tat-Trx1 reduced inflammatory damage by inhibiting inflammatory mediator and cytokine production. Collectively, these results demonstrated that Tat-Trx1 could exert anti-inflammatory effects by inhibiting the production of pro-inflammatory mediators and cytokines and by modulating MAPK signaling. Therefore, Tat-Trx1 may be a useful therapeutic agent for diseases induced by inflammatory damage.
Collapse
Affiliation(s)
- Eun Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Min Jea Shin
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Hyeon Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Yeon Joo Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Eun Jeong Sohn
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Lee Re Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, Gangwon 25457, Republic of Korea
| | - Hyun Ju Cha
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Sung Ho Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea.,Genesen Inc., Seoul 06181, Republic of Korea
| | - Sunghou Lee
- Department of Green Chemical Engineering, Sangmyung University, Cheonan, Chungcheongnam 31066, Republic of Korea
| | - Yeon Hee Yu
- Department of Anatomy and BK21 FOUR Project, College of Medicine, Soonchunhyang University, Cheonan, Chungcheongnam 31538, Republic of Korea
| | - Duk-Soo Kim
- Department of Anatomy and BK21 FOUR Project, College of Medicine, Soonchunhyang University, Cheonan, Chungcheongnam 31538, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, Gangwon 25457, Republic of Korea
| | - Jinseu Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Kyu Hyung Han
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Won Sik Eum
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| |
Collapse
|
14
|
Dey DK, Kang SC. CopA3 peptide induces permanent cell-cycle arrest in colorectal cancer cells. Mech Ageing Dev 2021; 196:111497. [PMID: 33957217 DOI: 10.1016/j.mad.2021.111497] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 12/23/2022]
Abstract
Cell-cycle arrest reflects an accumulation of responses to DNA damage that sequentially affects cell growth and division. Herein, we analyzed the effect of the 9-mer dimer defensin-like peptide, CopA3, against colorectal cancer cell growth and proliferation in a dose-dependent manner upon 96 h of treatment. As observed, CopA3 treatment significantly affected cancer cell growth, reduced colony formation ability, increased the number of SA-β-Gal positive cells, and remarkably reduced Ki67 protein expression. Notably, in HCT-116 cells, CopA3 (5 μM) treatment effectively increased oxidative stress and, as a result, amplified the endogenous ROS, mitochondrial ROS, and NO content in the cells, which further activated the DNA damage response and caused cell-cycle arrest at the G1 phase. The prolonged cell-cycle arrest elevated the release of inflammatory cytokines in the cell supernatant. Nevertheless, mechanistically, NAC treatment effectively reversed the CopA3 effect and significantly reduced the oxidative stress; subsequently rescuing the cells from G1 phase arrest. Overall, CopA3 treatment can inhibit the growth and proliferation of colorectal cancer cells by inducing cell-cycle arrest through the ROS-mediated pathway.
Collapse
Affiliation(s)
- Debasish Kumar Dey
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea.
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea.
| |
Collapse
|
15
|
Quan W, Kong S, Ouyang Q, Tao J, Lu S, Huang Y, Li S, Luo H. Use of 18β-glycyrrhetinic acid nanocrystals to enhance anti-inflammatory activity by improving topical delivery. Colloids Surf B Biointerfaces 2021; 205:111791. [PMID: 34022703 DOI: 10.1016/j.colsurfb.2021.111791] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/17/2021] [Accepted: 04/24/2021] [Indexed: 01/05/2023]
Abstract
18β-Glycyrrhetinic acid (GA) is often topically applied in clinical treatment of inflammatory skin diseases. However, GA has poor solubility in water, which results in poor skin permeability and low bioavailability. Nanocrystallization of drugs can enhance their permeability and improve bioavailability. We prepared GA nanocrystals (Nano GA) by high-pressure homogenization. These nanocrystals were characterized by photon correlation spectroscopy, scanning electron microscopy, thermogravimetric analysis, and X-ray diffractometry. The ability of Nano GA to improve dermal permeability was investigated ex vivo using Franz diffusion vertical cells and mouse skin. The topical anti-inflammatory activity of Nano GA was assessed in vivo by a 12-O-tetradecanoyl phorbol-13-acetate (TPA)-induced model in mouse ears. The average particle size of a GA nanocrystalline suspension was 288.6 ± 7.3 nm, with a narrow particle-size distribution (polydispersity index ∼0.13 ± 0.10), and the particle size of the lyophilized powder increased (552.0 ± 9.8 nm). After nanocrystallization, the thermal stability and crystallinity decreased but solubility increased significantly. Nano GA showed higher dermal permeability than Coarse GA. Macroscopic and staining-based observations of mouse ears and the levels of proinflammatory factors and myeloperoxidase revealed that the Nano GA hydrogel exhibited better anti-edema ability and more strongly inhibited inflammation development than the Coarse GA hydrogel and indomethacin hydrogel (positive drug). These results suggest that Nano GA could be an efficacious topical therapeutic agent for skin inflammation.
Collapse
Affiliation(s)
- Weiyan Quan
- Department of Applied Chemistry, School of Chemistry and Environmental Science, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Songzhi Kong
- Department of Applied Chemistry, School of Chemistry and Environmental Science, Guangdong Ocean University, Zhanjiang, 524088, China.
| | - Qianqian Ouyang
- The Marine Biomedical Research Institute of Guangdong Zhangjiang, Zhanjiang, 524023, China
| | - Jinlong Tao
- Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, 524001, China
| | - Sitong Lu
- Department of Applied Chemistry, School of Chemistry and Environmental Science, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Yongmei Huang
- The Marine Biomedical Research Institute of Guangdong Zhangjiang, Zhanjiang, 524023, China
| | - Sidong Li
- Department of Applied Chemistry, School of Chemistry and Environmental Science, Guangdong Ocean University, Zhanjiang, 524088, China; The Marine Biomedical Research Institute of Guangdong Zhangjiang, Zhanjiang, 524023, China
| | - Hui Luo
- The Marine Biomedical Research Institute of Guangdong Zhangjiang, Zhanjiang, 524023, China.
| |
Collapse
|
16
|
Chang SN, Khan I, Kim CG, Park SM, Choi DK, Lee H, Hwang BS, Kang SC, Park JG. Decursinol Angelate Arrest Melanoma Cell Proliferation by Initiating Cell Death and Tumor Shrinkage via Induction of Apoptosis. Int J Mol Sci 2021; 22:4096. [PMID: 33921050 PMCID: PMC8071397 DOI: 10.3390/ijms22084096] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
Melanoma is known to aggressively metastasize and is one of the prominent causes of skin cancer mortality. This study was designed to assess the molecular mechanism of decursinol angelate (DA) against murine melanoma cell line (B16F10 cells). Treatment of DA resulted in growth inhibition and cell cycle arrest at G0/G1 (p < 0.001) phase, evaluated through immunoblotting. Moreover, autophagy-related proteins such as ATG-5 (p < 0.0001), ATG-7 (p < 0.0001), beclin-1 (p < 0.0001) and transition of LC3-I to LC3-II (p < 0.0001) were markedly decreased, indicating autophagosome inhibition. Additionally, DA treatment triggered apoptotic events which were corroborated by the occurrence of distorted nuclei, elevated reactive oxygen species (ROS) levels and reduction in the mitochondrial membrane potential. Subsequently, there was an increase in the expression of pro-apoptotic protein Bax in a dose-dependent manner, with the corresponding downregulation of Bcl-2 expression and cytochrome C expression following 24 h DA treatment in A375.SM and B16F10 cells. We substantiated our results for apoptotic occurrence through flow cytometry in B16F10 cells. Furthermore, we treated B16F10 cells with N-acetyl-L-cysteine (NAC). NAC treatment upregulated ATG-5 (p < 0.0001), beclin-1 (p < 0.0001) and LC3-I to LC3-II (p < 0.0001) conversion, which was inhibited in the DA treatment group. We also noticed a systematic upregulation of important markers for progression of G1 cell phase such as CDK-2 (p < 0.029), CDK-4 (p < 0.036), cyclin D1 (p < 0.0003) and cyclin E (p < 0.020) upon NAC treatment. In addition, we also observed a significant fold reduction (p < 0.05) in ROS fluorescent intensity and the expression of Bax (p < 0.0001), cytochrome C (p < 0.0001), cleaved caspase-9 (p > 0.010) and cleaved caspase-3 (p < 0.0001). NAC treatment was able to ameliorate DA-induced apoptosis and cell cycle arrest to support our finding. Our in vivo xenograft model also revealed similar findings, such as downregulation of CDK-2 (p < 0.0001) and CDK-4 (p < 0.0142) and upregulation of Bax (p < 0.0001), cytochrome C (p < 0.0001), cleaved caspase 3 (p < 0.0001) and cleaved caspase 9 (p < 0.0001). In summary, our study revealed that DA is an effective treatment against B16F10 melanoma cells and xenograft mice model.
Collapse
Affiliation(s)
- Sukkum Ngullie Chang
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.N.C.); (C.G.K.); (S.M.P.)
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Korea;
| | - Imran Khan
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Korea;
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Chang Geon Kim
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.N.C.); (C.G.K.); (S.M.P.)
| | - Seon Min Park
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.N.C.); (C.G.K.); (S.M.P.)
| | - Dong Kyu Choi
- New Drug Development Center, DGMIF, 88 Dongnae-ro, Dong-gu, Daegu 41061, Korea; (D.K.C.); (H.L.)
| | - Heejin Lee
- New Drug Development Center, DGMIF, 88 Dongnae-ro, Dong-gu, Daegu 41061, Korea; (D.K.C.); (H.L.)
| | - Buyng Su Hwang
- Nakdonggang National Institute of Biological Resources, Sangju 37242, Korea;
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Korea;
| | - Jae Gyu Park
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.N.C.); (C.G.K.); (S.M.P.)
| |
Collapse
|
17
|
Conceição de Souza M, Hubner JT, Gonçalves GM, Ferreira da Costa Fernandes T, Laureano-Melo R, da Silva Côrtes W, Gonçalves L, de Assis da Silva F, Marinho BG. Topical and systemic use of Joannesia princeps vell. LC seed oil in acute pain and inflammation induced by different agents. JOURNAL OF ETHNOPHARMACOLOGY 2021; 268:113554. [PMID: 33152426 DOI: 10.1016/j.jep.2020.113554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Joannesia princeps (SOJP) has been used in folk medicine as anthelmintic treatment and cutaneous wound healing. AIM OF THE STUDY The purpose of this study is to evaluate the pharmacological activity of seed oil of Joannesia princeps, administered systemically and topically, on acute pain and inflammation. MATERIALS AND METHODS Male swiss mice were treated orally and topically with seed oil of Joannesia princeps in models of acute pain (acetic acid-induced abdominal writhing, formalin-induced licking behaviour and tail flick tests) and acute inflammation (carrageenan- and histamine-induced paw oedema; arachidonic acid-, capsaicin- and croton oil-induced ear oedema and air pouch tests), besides the open field model in the motor performance evaluation. RESULTS Seed oil of Joannesia princeps showed systemic action against acute pain in abdominal writhing test (37% and 56% inhibition in the number of writhes at doses of 30 and 100 mg/kg, respectively) and in the second phase of formalin-induced licking behaviour test (29%, 47 and 52% inhibition in the licking time at doses of 10, 30 and 100 mg/kg, respectively), as well as reducing croton oil-induced ear oedema by 72%, leukocyte recruitment and production of TNF-α and IL-6 in the air pouch tests. In addition, topical administration of SOJP inhibited carrageenan-induced paw oedema by 39% at dose of 500 μg/paw and inhibited histamine-induced oedema by 43 and 52% at doses of 300 and 500 μg/paw, respectively. SOJP also decreased croton oil-induced ear oedema by 67% at dose of 500 μg/paw and arachidonic acid-induced ear oedema by 63% at dose of 500 μg/paw, reducing the production of TNF-α, IL-1β and MIP2 in both. In addition, no adverse effects were observed at doses up to 2000 mg/kg. CONCLUSIONS Seed oil of Joannesia princeps presents antinociceptive and anti-inflammatory actions through its topical and systemic administration, promoted by inhibition of leukocyte recruitment and cytokine production (TNF-α, IL-1β, IL-6 and MIP-2).
Collapse
Affiliation(s)
- Milene Conceição de Souza
- Pharmacology Laboratory, Institute of Biological and Health Sciences, Rural Federal University of Rio de Janeiro, Seropédica, RJ, Brazil.
| | - Jhones Trindade Hubner
- Department of Chemistry, Rural Federal University of Rio de Janeiro, Seropédica, RJ, Brazil.
| | - Gabriela Mastrangelo Gonçalves
- Pharmacology Laboratory, Institute of Biological and Health Sciences, Rural Federal University of Rio de Janeiro, Seropédica, RJ, Brazil; Graduate Program in Physiological Sciences, Institute of Biological and Health Sciences, Rural Federal University of Rio de Janeiro, Seropédica, RJ, Brazil.
| | | | - Roberto Laureano-Melo
- Behavioral Physiopharmacology Laboratory, Barra Mansa Center University, Barra Mansa, RJ, Brazil.
| | - Wellington da Silva Côrtes
- Pharmacology Laboratory, Institute of Biological and Health Sciences, Rural Federal University of Rio de Janeiro, Seropédica, RJ, Brazil; Graduate Program in Physiological Sciences, Institute of Biological and Health Sciences, Rural Federal University of Rio de Janeiro, Seropédica, RJ, Brazil.
| | - Lenicio Gonçalves
- Department of Animal Biology, Rural Federal University of Rio de Janeiro, Seropédica, RJ, Brazil.
| | | | - Bruno Guimarães Marinho
- Pharmacology Laboratory, Institute of Biological and Health Sciences, Rural Federal University of Rio de Janeiro, Seropédica, RJ, Brazil; Graduate Program in Physiological Sciences, Institute of Biological and Health Sciences, Rural Federal University of Rio de Janeiro, Seropédica, RJ, Brazil.
| |
Collapse
|
18
|
Chang SN, Kim SH, Dey DK, Park SM, Nasif O, Bajpai VK, Kang SC, Lee J, Park JG. 5-O-Demethylnobiletin Alleviates CCl 4-Induced Acute Liver Injury by Equilibrating ROS-Mediated Apoptosis and Autophagy Induction. Int J Mol Sci 2021; 22:1083. [PMID: 33499185 PMCID: PMC7865239 DOI: 10.3390/ijms22031083] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 01/08/2023] Open
Abstract
Polymethoxyflavanoids (PMFs) have exhibited a vast array of therapeutic biological properties. 5-O-Demethylnobiletin (5-DN) is one such PMF having anti-inflammatory activity, yet its role in hepatoprotection has not been studied before. Results from in vitro study revealed that 5-DN did not exert a high level of cytotoxicity on HepG2 cells at 40 μM, and it was able to rescue HepG2 cell death induced by carbon tetrachloride (CCl4). Subsequently, we investigated acute liver injury on BALB/c mice induced by CCl4 through the intraperitoneal injection of 1 mL/kg CCl4 and co-administration of 5-DN at (1 and 2 mg/kg) by oral gavage for 15 days. The results illustrated that treatment with 5-DN attenuated CCl4-induced elevated serum aminotransferase (AST)/alanine aminotransferase (ALT) ratio and significantly ameliorated severe hepatic damage such as inflammation and fibrosis evidenced through lesser aberrations in the liver histology of 5-DN dose groups. Additionally, 5-DN efficiently counteracted and equilibrated the production of ROS accelerated by CCl4 and dramatically downregulated the expression of CYP2E1 vitally involved in converting CCl4 to toxic free radicals and also enhanced the antioxidant enzymes. 5-DN treatment also inhibited cell proliferation and inflammatory pathway abnormally regulated by CCl4 treatment. Furthermore, the apoptotic response induced by CCl4 treatment was remarkably reduced by enhanced Bcl-2 expression and noticeable reduction in Bax, Bid, cleaved caspase 3, caspase 9, and apaf-1 expression. 5-DN treatment also induced the conversion of LC3 and promoted the autophagic flux. Conclusively, 5-DN exhibited hepatoprotective effects in vitro and in vivo and prevented liver fibrosis induced by CCl4.
Collapse
Affiliation(s)
- Sukkum Ngullie Chang
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Korea; (S.N.C.); (D.K.D.); (S.C.K.)
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.H.K.); (S.M.P.)
| | - Se Ho Kim
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.H.K.); (S.M.P.)
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Korea
| | - Debasish Kumar Dey
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Korea; (S.N.C.); (D.K.D.); (S.C.K.)
| | - Seon Min Park
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.H.K.); (S.M.P.)
| | - Omaima Nasif
- Department of Physiology, College of Medicine, King Saud University (Medical City), King Khalid University Hospital, P.O. Box 2925, Riyadh 11461, Saudi Arabia;
| | - Vivek K. Bajpai
- Department of Energy and Materials Engineering, Dongguk University-Seoul, 30 Pildong-ro 1-gil, Seoul 04620, Korea
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Korea; (S.N.C.); (D.K.D.); (S.C.K.)
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Korea
| | - Jae Gyu Park
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.H.K.); (S.M.P.)
| |
Collapse
|
19
|
Chang SN, Dey DK, Oh ST, Kong WH, Cho KH, Al-Olayan EM, Hwang BS, Kang SC, Park JG. Phorbol 12-Myristate 13-Acetate Induced Toxicity Study and the Role of Tangeretin in Abrogating HIF-1α-NF-κB Crosstalk In Vitro and In Vivo. Int J Mol Sci 2020; 21:9261. [PMID: 33291656 PMCID: PMC7729754 DOI: 10.3390/ijms21239261] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/26/2020] [Accepted: 12/01/2020] [Indexed: 11/16/2022] Open
Abstract
Phorbol 12-myristate 13-acetate (PMA) is a potent tumor promoter and highly inflammatory in nature. Here, we investigated the toxic effects of PMA on different model system. PMA (10 μg) caused chromosomal aberrations on the Allium cepa root tip and induced mitotic dysfunction. Similarly, PMA caused embryonic and larval deformities and a plummeted survivability rate on zebrafish embryo in a dose-dependent manner. Persistently, PMA treatment on immortalized human keratinocyte human keratinocyte (HaCaT) cells caused massive inflammatory rush at 4 h and a drop in cell survivability at 24 h. Concomitantly, we replicated a cutaneous inflammation similar to human psoriasis induced by PMA. Herein, we used tangeretin (TAN), as an antagonist to counteract the inflammatory response. Results from an in vivo experiment indicated that TAN (10 and 30 mg/kg) significantly inhibited PMA stimulated epidermal hyperplasia and intra-epidermal neutrophilic abscesses. In addition, its treatment effectively neutralized PMA induced elevated reactive oxygen species (ROS) generation on in vitro and in vivo systems, promoting antioxidant response. The association of hypoxia-inducible factor 1-alpha (HIF-1α)-nuclear factor kappa-light-chain-enhancer of activated b cells (NF-κB) crosstalk triggered by PMA enhanced PKCα-ERK1/2-NF-κB pathway; its activation was also significantly counteracted after TAN treatment. Conclusively, we demonstrated TAN inhibited the nuclear translocation of HIF-1α and NF-κB p65. Collectively, TAN treatment ameliorated PMA incited malignant inflammatory response by remodeling the cutaneous microenvironment.
Collapse
Affiliation(s)
- Sukkum Ngullie Chang
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Korea; (S.N.C.); (D.K.D.)
- Advanced Bio Convergence Center, Pohang Technopark Foundation, Pohang 37668, Gyeongbuk, Korea; (S.T.O.); (W.H.K.)
| | - Debasish Kumar Dey
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Korea; (S.N.C.); (D.K.D.)
| | - Seong Taek Oh
- Advanced Bio Convergence Center, Pohang Technopark Foundation, Pohang 37668, Gyeongbuk, Korea; (S.T.O.); (W.H.K.)
- Okinawa Research Center Co. Ltd., 13-33, Suzaki, Uruma-si, Okinawa Ken 904-2234, Japan
| | - Won Ho Kong
- Advanced Bio Convergence Center, Pohang Technopark Foundation, Pohang 37668, Gyeongbuk, Korea; (S.T.O.); (W.H.K.)
| | - Kiu Hyung Cho
- Research Group, Gyeongbuk Institute for Bio Industry (GIB), Andong 36728, Korea;
| | - Ebtesam M. Al-Olayan
- Department of Zoology, Faculty of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Buyng Su Hwang
- Nakdonggang National Institute of Biological Resources, Sangju 37242, Korea;
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Korea; (S.N.C.); (D.K.D.)
| | - Jae Gyu Park
- Advanced Bio Convergence Center, Pohang Technopark Foundation, Pohang 37668, Gyeongbuk, Korea; (S.T.O.); (W.H.K.)
| |
Collapse
|
20
|
Wang L, Lee W, Jayawardena TU, Cha SH, Jeon YJ. Dieckol, an algae-derived phenolic compound, suppresses airborne particulate matter-induced skin aging by inhibiting the expressions of pro-inflammatory cytokines and matrix metalloproteinases through regulating NF-κB, AP-1, and MAPKs signaling pathways. Food Chem Toxicol 2020; 146:111823. [PMID: 33164846 DOI: 10.1016/j.fct.2020.111823] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023]
Abstract
Exposure to particulate matter causes skin aging. In the present study, we investigated the effect of an algae-derived phenolic compound, dieckol (DK), against Chinese particulate matter (CPM)-stimulated aging in vitro in human dermal fibroblasts (HDF cells) and in vivo in zebrafish. DK effectively protected HDF cells against CPM-induced oxidative stress by scavenging intracellular reactive oxygen species. Moreover, DK significantly improved collagen synthesis and inhibited intracellular collagenase activity in CPM-stimulated HDF cells. In addition, DK remarkably reduced the expression of pro-inflammatory cytokines and matrix metalloproteinases via regulating the nuclear factor kappa B, activator protein 1, and mitogen-activated protein kinases signaling pathways in CPM-stimulated HDF cells. Furthermore, the in vivo test results demonstrated that DK effectively improved the survival rate of CPM-stimulated zebrafish via suppressing oxidative stress and inflammatory response. In conclusion, this study suggests that DK is a potential anti-aging compound that can be used as a therapeutic agent to improve CPM-induced skin aging, or as an ingredient to develop a cosmetic or medicine in the cosmeceutical and pharmaceutical industries.
Collapse
Affiliation(s)
- Lei Wang
- Department of Marine Life Sciences, Jeju National University, Jeju, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea
| | - WonWoo Lee
- Freshwater Bioresources Utilization Division, Nakdonggang National Institute of Biological Resources, Sangju, 37242, Republic of Korea
| | - Thilina U Jayawardena
- Department of Marine Life Sciences, Jeju National University, Jeju, 63243, Republic of Korea
| | - Seon-Heui Cha
- Department of Marine Bio and Medical Science, Hanseo University, Chungcheognam-do, 32158, Republic of Korea
| | - You-Jin Jeon
- Department of Marine Life Sciences, Jeju National University, Jeju, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea.
| |
Collapse
|
21
|
Dey DK, Kang SC. Aflatoxin B1 induces reactive oxygen species-dependent caspase-mediated apoptosis in normal human cells, inhibits Allium cepa root cell division, and triggers inflammatory response in zebrafish larvae. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 737:139704. [PMID: 32512299 DOI: 10.1016/j.scitotenv.2020.139704] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/23/2020] [Accepted: 05/23/2020] [Indexed: 06/11/2023]
Abstract
Mycotoxin contamination of food and water is a serious global concern. Aflatoxin B1 (AFB1) is a deadly mycotoxin that contaminates both food and water bodies in the environment. AFB1 is reported to cause severe health issues, including hepatotoxicity, teratogenicity, and immunotoxicity in humans; however, the mechanistic effects on plant and aquatic animals are not fully understood. To obtain a clear understanding of the effects of AFB1 on the ecosystem, we examined the influence of AFB1 exposure on different model systems corresponding to various habitats. In the current study, AFB1 contamination consequences were studied on a human normal cell lines (HaCaT, CCD 841 CoN), meristematic Allium cepa (onion) root cells, and zebrafish embryonic development. Our results clearly indicate that concentrations of AFB1 >10 μM are toxic to HaCaT cells. Morphological changes of HaCaT and CCD 841 CoN cells were clearly observed after exposure to AFB1. Particularly in HaCaT cells, treatment with 50 μM and 100 μM AFB1induces oxidative stress by excessive endogenous free-radical production such as ROS and NO generation. These consequences accelerate the ROS-dependent DNA damage events, which subsequently result in caspase mediated programmed cell death. Exposure of A. cepa root cells to AFB1 for 24 h resulted in abnormal cell division. A. cepa root cells subjected to AFB1 treatment showed a significant concentration-dependent increase in metaphase arrest. Exposure of zebrafish embryos to AFB1 also revealed that AFB1 contamination restricts the larval growth and development, resulting in a remarkably increased zebrafish mortality rate. Collectively, results of the current study indicate that AFB1 contamination triggers the programmed cell death machinery, subsequently affecting the ecosystem.
Collapse
Affiliation(s)
- Debasish Kumar Dey
- Department of Biotechnology, Daegu University, Jillyang, Naeri-ri, Gyeongsan, Gyeongbuk 38453, Republic of Korea.
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Jillyang, Naeri-ri, Gyeongsan, Gyeongbuk 38453, Republic of Korea.
| |
Collapse
|
22
|
Lee SE, Kim JH, Lim C, Cho S. Neuroprotective effect of Angelica gigas root in a mouse model of ischemic brain injury through MAPK signaling pathway regulation. Chin Med 2020; 15:101. [PMID: 32983252 PMCID: PMC7509924 DOI: 10.1186/s13020-020-00383-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/18/2020] [Indexed: 11/10/2022] Open
Abstract
Background The root of Angelica gigas Nakai (Apiaceae) has been traditionally used as an important herbal medicine to treat blood-deficiency-related disorders in Eastern Asian countries, and recently, it has been recognized as a potential candidate for improving cardiovascular diseases. Methods In this study, the neuroprotective effect of a methanol extract of A. gigas root (RAGE) was investigated in a mouse stroke model induced by a 90 min transient middle cerebral artery occlusion (tMCAO). Infarction volumes and morphological changes in brain tissues were measured using TTC, cresyl violet, and H&E staining. The neuroprotective mechanism of RAGE was elucidated through investigation of protein expression levels using western blotting, IHC, and ELISA assays. The plasma concentrations of decursin, a major compound in RAGE, were measured after oral administration of RAGE to SD rats. Results The infarction volumes in brain tissues were significantly reduced and the morphological deteriorations in the brain neuron cells were improved in tMCAO mice when pre-treated with RAGE at 1000 mg/(kg bw·d) for two consecutive days. The neuroprotective mechanism of RAGE was confirmed to attenuate ERK-related MAPK signaling pathways in the ipsilateral hippocampus hemisphere in mice. The concentrations of decursin in rat plasma samples showed peak absorption and elimination in vivo after oral administration of RAGE at 100 mg/rat. Conclusion Mice administered RAGE before the tMCAO operation had less neuronal cell death than those that were not administered RAGE prior to the operation, and this study provides preclinical evidence for use of A. gigas in ischemic stroke.
Collapse
Affiliation(s)
- Se-Eun Lee
- Department of Korean Medicine, School of Korean Medicine, Yangsan Campus of Pusan National University, Yangsan, 50612 Republic of Korea
| | - Jung-Hoon Kim
- Department of Korean Medicine, School of Korean Medicine, Yangsan Campus of Pusan National University, Yangsan, 50612 Republic of Korea
| | - Chiyeon Lim
- Department of Medicine, College of Medicine, Dongguk University, Goyang, 10326 Republic of Korea
| | - Suin Cho
- Department of Korean Medicine, School of Korean Medicine, Yangsan Campus of Pusan National University, Yangsan, 50612 Republic of Korea
| |
Collapse
|
23
|
Dey DK, Chang SN, Vadlamudi Y, Park JG, Kang SC. Synergistic therapy with tangeretin and 5-fluorouracil accelerates the ROS/JNK mediated apoptotic pathway in human colorectal cancer cell. Food Chem Toxicol 2020; 143:111529. [PMID: 32619557 DOI: 10.1016/j.fct.2020.111529] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/05/2020] [Accepted: 06/12/2020] [Indexed: 12/24/2022]
Abstract
Synergistic therapy is emerging as a promising strategy for improving the chemotherapeutic efficacy of anticancer drugs. Addition of adjuvants with standard anticancer drugs has shown successful reduction of adverse side effects. The synthetic drug 5-Fluorouracil (5-FU) shows several side effects upon prolonged chemotherapy, thereby restricting its long-term clinical application. Several studies have reported anticancer potential and anti-inflammatory activity of tangeretin (TAN) towards mammalian cells. Therefore, we investigate whether the combination of TAN with 5-FU increases their anticancer potential against colorectal cancer. In this study, we examined the synergistic activity of TAN and 5-FU on the viability of several human cancer and normal cells. Several possible mechanistic pathways were screened, and found that co-exposure of TAN and 5-FU accelerates oxidative-stress and increases endogenous-ROS generation, which sequentially triggers the DNA damage response and activates the apoptotic pathway, by down-regulating autophagy and DNA repair system in HCT-116 cells. TAN and 5-FU co-treatment also remarkably reduces the mitochondrial membrane potential, and sequentially decreases ATPase activity. Collectively, results indicate that combination of TAN and 5-FU significantly accelerates apoptosis via JNK mediated pathway. To our knowledge gained from literature, this study is the first to describe synergistic activity of TAN and 5-FU against colorectal cancer cells.
Collapse
Affiliation(s)
- Debasish Kumar Dey
- Department of Biotechnology, Daegu University, Gyeongsan, 38453, Republic of Korea
| | - Sukkum Ngullie Chang
- Department of Biotechnology, Daegu University, Gyeongsan, 38453, Republic of Korea
| | | | - Jae Gyu Park
- Advanced Bio Convergence Center, Pohang Technopark Foundation, Pohang, Gyeongbuk, 37668, Republic of Korea.
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, 38453, Republic of Korea.
| |
Collapse
|
24
|
Lee TK, Kim B, Kim DW, Ahn JH, Sim H, Lee JC, Yang GE, Her Y, Park JH, Kim HS, Sim TH, Lee HS, Won MH. Effects of Decursin and Angelica gigas Nakai Root Extract on Hair Growth in Mouse Dorsal Skin via Regulating Inflammatory Cytokines. Molecules 2020; 25:E3697. [PMID: 32823713 PMCID: PMC7464339 DOI: 10.3390/molecules25163697] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/06/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023] Open
Abstract
This current study investigates the facilitative effects and mechanisms of decursin, a major component of Angelica gigas Nakai (AGN), and AGN root extract on hair growth in mice. We perform high-performance liquid chromatography on AGN extract to show it contains 7.3% decursin. Hairs in mouse dorsal skin are shaved distilled in water, 0.15% decursin, and 2% AGN root extract (0.15% decursin in the diluted extract) and topically applied twice a day for 17 days. Hematoxylin and eosin staining are done to examine the morphological changes in the hair follicles. To compare the effects of decursin and AGN extract on inflammatory cytokines in the dorsal skin, Western blot analysis and immunohistochemistry for tumor necrosis factor α (TNF-α) and interleukin (IL)-1β as pro-inflammatory cytokines, and IL-4 and IL-13 as anti-inflammatory cytokines are conducted. The results show that the application of decursin and AGN extract confer effects on hair growth. Hair growth is significantly facilitated from seven days after the treatments compared to that in the control group, and completely grown hair was found 17 days after the treatments. The protein levels and immunoreactivity of TNF-α and IL-1β in this case are significantly decreased, whereas the IL-4 and IL-13 levels and immunoreactivity are significantly increased compared to those in the control group. Additionally, high-mobility group box 1, an inflammatory mediator, is elevated by the topical application of decursin and AGN extract. Taken together, the treatment of mouse dorsal skin with AGE root extract containing decursin promotes hair growth by regulating pro- and/or anti-inflammatory cytokines. We, therefore, suggest that AGN root extract as well as decursin can be utilized as materials for developing hair growth-facilitating treatments.
Collapse
Affiliation(s)
- Tae-Kyeong Lee
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Korea; (T.-K.L.); (J.H.A.)
| | - Bora Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Korea; (B.K.); (H.S.); (J.-C.L.)
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, and Research Institute of Oral Sciences, College of Dentistry, Gangnung-Wonju National University, Gangneung, Gangwon 25457, Korea;
| | - Ji Hyeon Ahn
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Korea; (T.-K.L.); (J.H.A.)
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Korea; (B.K.); (H.S.); (J.-C.L.)
| | - Hyejin Sim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Korea; (B.K.); (H.S.); (J.-C.L.)
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Korea; (B.K.); (H.S.); (J.-C.L.)
| | - Go Eun Yang
- Department of Radiology, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Gangwon 24289, Korea;
| | - Young Her
- Department of Dermatology, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Gangwon 24289, Korea;
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju, Gyeongbuk 38066, Korea;
| | - Hyun Sook Kim
- Leefarm Co., Ltd., Hongcheon, Gangwon 25117, Korea; (H.S.K.); (T.H.S.)
| | - Tae Heung Sim
- Leefarm Co., Ltd., Hongcheon, Gangwon 25117, Korea; (H.S.K.); (T.H.S.)
| | - Hyun Sam Lee
- Leefarm Co., Ltd., Hongcheon, Gangwon 25117, Korea; (H.S.K.); (T.H.S.)
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Korea; (B.K.); (H.S.); (J.-C.L.)
| |
Collapse
|
25
|
Dey DK, Kang SC. Weissella confusa DD_A7 pre-treatment to zebrafish larvae ameliorates the inflammation response against Escherichia coli O157:H7. Microbiol Res 2020; 237:126489. [PMID: 32464536 DOI: 10.1016/j.micres.2020.126489] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/14/2020] [Accepted: 04/25/2020] [Indexed: 11/17/2022]
Abstract
Increasing multidrug-resistant pathogenic bacterial contamination in the environment has become the leading cause of food poisoning, resulting in life-threatening conditions due to late detection and limited therapeutic options. Escherichia coli O157:H7 is one such pathogen which is severely affecting the environmental livestock and ultimately leads to human infection. In this context, probiotics could be a useful strategy to minimize the growth of pathogens, as they produce several antimicrobial compounds and shows an exclusive competitive behavior against the pathogens. Therefore, supplementation of probiotics is wieldy accepted in the field of agriculture for the maintenance of animal's health. Previously, we reported that W. confusa DD_A7 possesses anti-bacterial and immune-stimulatory activity in-vitro. Therefore, in the present study, we investigated the impact of oral-administration of DD_A7 powder against E. coli O157:H7. The 6 days post-fertilized zebrafish larvae were used to evaluate the pathogenicity of the microbe. 1 × 108 CFU/ml of E. coli O157:H7 effectively induced the inflammatory response in zebrafish larvae. Where 1 × 108 CFU/ml DD_A7 pre-treatment prolonged the survivability of zebrafish larvae and improved the immune response of zebrafish larvae against pathogenic infection. The antibacterial property of DD_A7 against the pathogen correlated with the significant reduction of oxidative stress and host inflammatory response, by inhibiting NF-κB and its downstream signaling pathway. The findings demonstrated the prophylactic activity of DD_A7 suggesting that its supplementation improved the host defense mechanism by reducing oxidative stress. The growth of pathogen was effectively suppressed in the DD_A7 pre-treated larvae and maintained a healthy gastrointestinal environment in the zebrafish model.
Collapse
Affiliation(s)
- Debasish Kumar Dey
- Department of Biotechnology, Daegu University, Jillyang, Naeri-ri, Gyeongsan, Gyeongbuk, 38453, Republic of Korea.
| | - Sun Chul Kang
- Department of Biotechnology, College of Engineering, Daegu University, Jillyang, Naeri-ri, Gyeongsan, Gyeongbuk, 38453, Republic of Korea.
| |
Collapse
|
26
|
Khan I, Dey DK, Lee JH, Kang SC. Bamboo leave extract ameliorated 12-O-tetradecanoylphorbol-13-acetate (TPA) induced ear inflammation by reducing MAP kinase levels and NF-κB activation in mice model. Nat Prod Res 2020; 35:5299-5303. [PMID: 32290690 DOI: 10.1080/14786419.2020.1753053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Sasa coreana Nakai (SCN) is a medicinal plant commonly used against inflammation. However, the underlined mechanisms against skin inflammation is poorly understood. The present study investigated the effects of SCN leave extract on ear inflammation. To this aim, six-week-old male ICR mice was subjected to 12-O-tetradecanoyl-phorbol-13-acetate induce ear edema, which were then topically treated with the leave extract of SCN. Ear thickness, weight, and morphological changes were recorded to ensure the induction of ear edema. Further, histological analysis and protein expression for inflammatory markers were also recorded to validate the study. Topical treatment with SCN repressed TPA-induced ear edema in a dose-dependent manner. Further, SCN treatment significantly antagonized the protein expression of MAP kinase signaling pathway and reduced the effect of TPA-induced NF-κB activation, sequentially, deactivated its transcriptional targets in a dose-dependent manner. Collectively, the study suggested that SCN could be a useful therapeutic agent against skin inflammation.
Collapse
Affiliation(s)
- Imran Khan
- Department of Biotechnology, Daegu University, Gyeongsan, Republic of Korea
| | - Debasish Kumar Dey
- Department of Biotechnology, Daegu University, Gyeongsan, Republic of Korea
| | - Jai-Heon Lee
- Department of Genetic Engineering, Dong-A University, Busan, Republic of Korea
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Republic of Korea
| |
Collapse
|
27
|
Jung HG, Kim HH, Dey DK, Yu JM, Kim H, Cho YH, Jang JY, Park JY, Kang SC, An BJ. The Anti-inflammatory and Immune-Boosting Potential of Quercetin-3-O-β-D-glucopyranosyl-(1 → 6)-β-D-glucopyranoside in LPS–Stimulated RAW264.7 Macrophages. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s43450-020-00027-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|